Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 28(23): 5030-5039, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-35579997

RESUMEN

Immunotherapy targeting coinhibitory receptors has been highly successful in treating a wide variety of malignancies; however, only a subset of patients exhibits durable responses. The first FDA-approved immunotherapeutics targeting coinhibitory receptors PD1 and CTLA4, alone or in combination, significantly improved survival but were also accompanied by substantial toxicity in combination. The third FDA-approved immune checkpoint inhibitor targets LAG3, a coinhibitory receptor expressed on activated CD4+ and CD8+ T cells, especially in settings of long-term antigenic stimulation, such as chronic viral infection or cancer. Mechanistically, LAG3 expression limits both the expansion of activated T cells and the size of the memory pool, suggesting that LAG3 may be a promising target for immunotherapy. Importantly, the mechanism(s) by which LAG3 contributes to CD8+ T-cell exhaustion may be distinct from those governed by PD1, indicating that the combination of anti-LAG3 and anti-PD1 may synergistically enhance antitumor immunity. Clinical studies evaluating the role of anti-LAG3 in combination with anti-PD1 are underway, and recent phase III trial results in metastatic melanoma demonstrate both the efficacy and safety of this combination. Further ongoing clinical trials are evaluating this combination across multiple tumor types and the adjuvant setting, with accompanying translational and biomarker-focused studies designed to elucidate the molecular pathways that lead to improved antitumor T-cell responses following dual blockade of PD1 and LAG3. Overall, LAG3 plays an important role in limiting T-cell activation and has now become part of the repertoire of combinatorial immunotherapeutics available for the treatment of metastatic melanoma.


Asunto(s)
Melanoma , Receptor de Muerte Celular Programada 1 , Humanos , Antígenos CD/metabolismo , Linfocitos T CD8-positivos , Inmunoterapia/métodos , Melanoma/tratamiento farmacológico , Melanoma/genética , Ensayos Clínicos Fase III como Asunto
2.
Cell Rep Med ; 2(12): 100476, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34873589

RESUMEN

Despite extensive analyses, there remains an urgent need to delineate immune cell states that contribute to mortality in people critically ill with COVID-19. Here, we present high-dimensional profiling of blood and respiratory samples from people with severe COVID-19 to examine the association between cell-linked molecular features and mortality outcomes. Peripheral transcriptional profiles by single-cell RNA sequencing (RNA-seq)-based deconvolution of immune states are associated with COVID-19 mortality. Further, persistently high levels of an interferon signaling module in monocytes over time lead to subsequent concerted upregulation of inflammatory cytokines. SARS-CoV-2-infected myeloid cells in the lower respiratory tract upregulate CXCL10, leading to a higher risk of death. Our analysis suggests a pivotal role for viral-infected myeloid cells and protracted interferon signaling in severe COVID-19.


Asunto(s)
COVID-19/inmunología , COVID-19/mortalidad , Pulmón/inmunología , SARS-CoV-2/patogenicidad , Anciano , COVID-19/sangre , COVID-19/virología , Enfermedad Crítica , Citocinas/sangre , Redes Reguladoras de Genes , Humanos , Inflamación , Pulmón/virología , Modelos Teóricos , Monocitos/inmunología , Células Mieloides/inmunología , Reproducibilidad de los Resultados , Carga Viral
3.
Immunity ; 54(10): 2209-2217.e6, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34551314

RESUMEN

CD4+ T cells share common developmental pathways with CD8+ T cells, and upon maturation, CD4+ T conventional T (Tconv) cells lack phenotypic markers that distinguish these cells from FoxP3+ T regulatory cells. We developed a tamoxifen-inducible ThPOKCreERT2.hCD2 line with Frt sites inserted on either side of the CreERT2-hCD2 cassette, and a Foxp3Ametrine-FlpO strain, expressing Ametrine and FlpO in Foxp3+ cells. Breeding these mice resulted in a CD4conviCreERT2-hCD2 line that allows for the specific manipulation of a gene in CD4+ Tconv cells. As FlpO removes the CreERT2-hCD2 cassette, CD4+ Treg cells are spared from Cre activity, which we refer to as allele conditioning. Comparison with an E8IiCreERT2.GFP mouse that enables inducible targeting of CD8+ T cells, and deletion of two inhibitory receptors, PD-1 and LAG-3, in a melanoma model, support the fidelity of these lines. These engineered mouse strains present a resource for the temporal manipulation of genes in CD4+ T cells and CD4+ Tconv cells.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Edición Génica/métodos , Integrasas/genética , Alelos , Animales , Linfocitos T CD8-positivos/citología , Línea Celular , Ratones
4.
Sci Immunol ; 5(49)2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680952

RESUMEN

Mechanisms of resistance to cancer immunotherapy remain poorly understood. Lymphocyte activation gene-3 (LAG3) signaling is regulated by a disintegrin and metalloprotease domain-containing protein-10 (ADAM10)- and ADAM17-mediated cell surface shedding. Here, we show that mice expressing a metalloprotease-resistant, noncleavable LAG3 mutant (LAG3NC) are resistant to PD1 blockade and fail to mount an effective antitumor immune response. Expression of LAG3NC intrinsically perturbs CD4+ T conventional cells (Tconvs), limiting their capacity to provide CD8+ T cell help. Furthermore, the translational relevance for these observations is highlighted with an inverse correlation between high LAG3 and low ADAM10 expression on CD4+ Tconvs in the peripheral blood of patients with head and neck squamous cell carcinoma, which corresponded with poor prognosis. This correlation was also observed in a cohort of patients with skin cancers and was associated with increased disease progression after standard-of-care immunotherapy. These data suggest that subtle changes in LAG3 inhibitory receptor signaling can act as a resistance mechanism with a substantive effect on patient responsiveness to immunotherapy.


Asunto(s)
Antígenos CD/inmunología , Resistencia a Antineoplásicos/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/inmunología , Proteína ADAM10/antagonistas & inhibidores , Proteína ADAM10/inmunología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Antígenos CD/sangre , Antígenos CD/genética , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunoterapia , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/inmunología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Transcriptoma , Proteína del Gen 3 de Activación de Linfocitos
5.
Nat Immunol ; 20(11): 1425-1434, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31611702

RESUMEN

Although immunotherapeutics targeting the inhibitory receptors (IRs) CTLA-4, PD-1 or PD-L1 have made substantial clinical progress in cancer, a considerable proportion of patients remain unresponsive to treatment. Targeting novel IR-ligand pathways in combination with current immunotherapies may improve clinical outcomes. New clinical immunotherapeutics target T cell-expressed IRs (LAG-3, TIM-3 and TIGIT) as well as inhibitory ligands in the B7 family (B7-H3, B7-H4 and B7-H5), although many of these targets have complex biologies and unclear mechanisms of action. With only modest clinical success in targeting these IRs, current immunotherapeutic design may not be optimal. This Review covers the biology of targeting novel IR-ligand pathways and the current clinical status of their immunotherapeutics, either as monotherapy or in combination with antibody to PD-1 or to its ligand PD-L1. Further understanding of the basic biology of these targets is imperative to the development of effective cancer immunotherapies.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Antígenos B7/antagonistas & inhibidores , Receptores Coestimuladores e Inhibidores de Linfocitos T/antagonistas & inhibidores , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Antígenos CD/inmunología , Enfermedades Autoinmunes/inmunología , Antígenos B7/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Quimioterapia Combinada/métodos , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Humanos , Ligandos , Terapia Molecular Dirigida/métodos , Neoplasias/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Proteína del Gen 3 de Activación de Linfocitos
6.
Immunology ; 157(3): 232-247, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31087644

RESUMEN

Regulatory T (Treg) cells play a crucial role in maintaining self-tolerance and resolution of immune responses by employing multifaceted immunoregulatory mechanisms. However, Treg cells readily infiltrate into the tumor microenvironment (TME) and dampen anti-tumor immune responses, thereby becoming a barrier to effective cancer immunotherapy. There has been a substantial expansion in the development of novel immunotherapies targeting various inhibitory receptors (IRs), such as CTLA4, PD1 and LAG3, but these approaches have mechanistically focused on the elicitation of anti-tumor responses. However, enhanced inflammation in the TME could also play a detrimental role by facilitating the recruitment, stability and function of Treg cells by up-regulating chemokines that promote Treg cell migration, and/or increasing inhibitory cytokine production. Furthermore, IR blockade may enhance Treg cell function and survival, thereby serving as a resistance mechanism against effective immunotherapy. Given that Treg cells are comprised of functionally and phenotypically heterogeneous sub-populations that may alter their characteristics in a context-dependent manner, it is critical to identify unique molecular pathways that are preferentially used by intratumoral Treg cells. In this review, we discuss markers that serve to identify certain Treg cell subsets, distinguished by chemokine receptors, IRs and cytokines that facilitate their migration, stability and function in the TME. We also discuss how these Treg cell subsets correlate with the clinical outcome of patients with various types of cancer and how they may serve as potential TME-specific targets for novel cancer immunotherapies.


Asunto(s)
Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Escape del Tumor , Animales , Quimiocinas/inmunología , Quimiocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Neoplasias/metabolismo , Neoplasias/patología , Fenotipo , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Microambiente Tumoral
7.
J Control Release ; 258: 208-217, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28501670

RESUMEN

Dry eye disease (DED) is a common ocular disorder affecting millions of individuals worldwide. The pathology of DED involves the infiltration of CD4+ lymphocytes, leading to tear film instability and destructive inflammation. In the healthy steady state, a population of immunosuppressive T-cells called regulatory T-cells (Treg) regulates proliferation of immune cells that would otherwise lead to a disruption of immunological homeostasis. For this reason, it has been suggested that Tregs could restore the immunological imbalance in DED. To this end, one possible approach would be to recruit the body's own, endogenous Tregs in order to enrich them at the site of inflammation and tissue destruction. Previously, we have demonstrated a reduction of inflammation and disease symptoms in models of periodontitis corresponding to recruitment of endogenous Tregs, which was accomplished by local placement of controlled release systems that sustain a gradient of the chemokine CCL22, referred to here as Treg-recruiting microspheres. Given that DED is characterized by a pro-inflammatory environment resulting in local tissue destruction, we hypothesized that the controlled release of CCL22 could also recruit Tregs to the ocular surface potentially mediating inflammation and symptoms of DED. Indeed, data suggest that Treg-recruiting microspheres are capable of overcoming the immunological imbalance of Tregs and CD4+ IFN-γ+ cells in the lacrimal gland. Administration of Treg-recruiting microspheres effectively mitigated the symptoms of DED as measured through a number of outcomes such as tear clearance, goblet cells density and corneal epithelial integrity, suggesting that recruitment of endogenous Treg can mitigate inflammation associated with DED.


Asunto(s)
Antiinflamatorios/administración & dosificación , Quimiocina CCL22/administración & dosificación , Preparaciones de Acción Retardada/química , Síndromes de Ojo Seco/complicaciones , Inflamación/prevención & control , Linfocitos T Reguladores/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Quimiocina CCL22/uso terapéutico , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Síndromes de Ojo Seco/inmunología , Femenino , Inflamación/inmunología , Ratones Endogámicos BALB C , Linfocitos T Reguladores/inmunología
8.
Immunol Rev ; 276(1): 80-96, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28258692

RESUMEN

Despite the impressive impact of CTLA4 and PD1-PDL1-targeted cancer immunotherapy, a large proportion of patients with many tumor types fail to respond. Consequently, the focus has shifted to targeting alternative inhibitory receptors (IRs) and suppressive mechanisms within the tumor microenvironment. Lymphocyte activation gene-3 (LAG3) (CD223) is the third IR to be targeted in the clinic, consequently garnering considerable interest and scrutiny. LAG3 upregulation is required to control overt activation and prevent the onset of autoimmunity. However, persistent antigen exposure in the tumor microenvironment results in sustained LAG3 expression, contributing to a state of exhaustion manifest in impaired proliferation and cytokine production. The exact signaling mechanisms downstream of LAG3 and interplay with other IRs remain largely unknown. However, the striking synergy between LAG3 and PD1 observed in multiple settings, coupled with the contrasting intracellular cytoplasmic domain of LAG3 as compared with other IRs, highlights the potential uniqueness of LAG3. There are now four LAG3-targeted therapies in the clinic with many more in preclinical development, emphasizing the broad interest in this IR. Given the translational relevance of LAG3 and the heightened interest in the impact of dual LAG3/PD1 targeting in the clinic, the outcome of these trials could serve as a nexus; significantly increasing or dampening enthusiasm for subsequent targets in the cancer immunotherapeutic pipeline.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Activación de Linfocitos , Ratones , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal , Escape del Tumor , Microambiente Tumoral , Proteína del Gen 3 de Activación de Linfocitos
9.
Immunity ; 44(2): 316-29, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26872697

RESUMEN

Regulatory T (Treg) cells pose a major barrier to effective anti-tumor immunity. Although Treg cell depletion enhances tumor rejection, the ensuing autoimmune sequelae limits its utility in the clinic and highlights the need for limiting Treg cell activity within the tumor microenvironment. Interleukin-35 (IL-35) is a Treg cell-secreted cytokine that inhibits T cell proliferation and function. Using an IL-35 reporter mouse, we observed substantial enrichment of IL-35(+) Treg cells in tumors. Neutralization with an IL-35-specific antibody or Treg cell-restricted deletion of IL-35 production limited tumor growth in multiple murine models of human cancer. Limiting intratumoral IL-35 enhanced T cell proliferation, effector function, antigen-specific responses, and long-term T cell memory. Treg cell-derived IL-35 promoted the expression of multiple inhibitory receptors (PD1, TIM3, LAG3), thereby facilitating intratumoral T cell exhaustion. These findings reveal previously unappreciated roles for IL-35 in limiting anti-tumor immunity and contributing to T cell dysfunction in the tumor microenvironment.


Asunto(s)
Anticuerpos Bloqueadores/administración & dosificación , Interleucinas/metabolismo , Melanoma Experimental/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Memoria Inmunológica , Interleucinas/genética , Interleucinas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Microambiente Tumoral , Proteína del Gen 3 de Activación de Linfocitos
10.
Eur J Immunol ; 45(7): 1892-905, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26018646

RESUMEN

Inhibitory receptors expressed on T cells control immune responses while limiting autoimmunity. However, tumors can hijack these "checkpoints" for protection from immune attack. Tumor-specific T cells that exhibit an exhausted, unresponsive phenotype express high levels of inhibitory receptors including CTLA4, PD1, and LAG3, among others. Intratumoral regulatory T cells promote immunosuppression and also express multiple inhibitory receptors. Overcoming this inhibitory receptor-mediated immune tolerance has thus been a major focus of recent cancer immunotherapeutic developments. Here, we review how boosting the host's immune system by blocking inhibitory receptor signaling with antagonistic mAbs restores the capacity of T cells to drive durable antitumor immune responses. Clinical trials targeting the CTLA4 and PD1 pathways have shown durable effects in multiple tumor types. Many combinatorial therapies are currently being investigated with encouraging results that highlight enhanced antitumor immunogenicity and improved patient survival. Finally, we will discuss the ongoing identification and dissection of novel T-cell inhibitory receptor pathways, which could lead to the development of new combinatorial therapeutic approaches.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Escape del Tumor/inmunología , Animales , Antígenos CD/inmunología , Antígeno CTLA-4/inmunología , Humanos , Receptor de Muerte Celular Programada 1/inmunología , Proteína del Gen 3 de Activación de Linfocitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...